Cómo citar
Osorio, J. H. . (2006). Variantes genéticas y metabolismo de los opiáceos, los opioides y la cocaína. Cultura Y Droga, 11(13), 39–54. Recuperado a partir de https://revistasojs.ucaldas.edu.co/index.php/culturaydroga/article/view/5983

Autores/as

José Henry Osorio
josheno@yahoo.com

Resumen

Resumen: Algunas variantes en los genes que codifican proteínas involucradas en el metabolismo o biotransformación de drogas de abuso, pueden afectar la vulnerabilidad a desarrollar una adicción; y otras variantes relacionadas con el metabolismo de los opiáceos y la cocaína, han sido identificadas y pueden estar asociadas con la vulnerabilidad a desarrollar o afectar el tratamiento de las enfermedades adictivas. Esta revisión está enfocada sobre los opiáceos, los opioides y la cocaína, mencionando algunas variantes genéticas que pueden alterar su metabolismo.

Arnsten A.F. (2000a). Through the looking glass: differential noradrenergic modulation of prefrontal cortical function. Neural Plast 7: 133-146.

_________. (2000b). Stress impairs prefrontal cortical function in rats and monkeys: role of dopamine D1 and norepinephrine-1 receptor mechanisms. Prog Brain Res 126: 183-192.

Azaryan AV, Coughlin LJ. Buzas B, Clock BJ. and Cox BM (1996) Effect of chronic cocaine treatment on- and-opioid receptor mRNA levels in dopaminergically innervated brain regions. J Neurochem 66: 443-448.

Bart G. Borg L. Schluger J.H. Green M. Ho A. and Kreek M.J. (2003). Suppressed prolactin response to dynorphin A (1-13) in methadone maintained versus control subjects. J Pharmacol Exp Ther 306: 581-587.

Becker A. Grecksch G. Brodemann R. Kraus J. Peters B. Schroeder H. Thiemann W. Loh H.H. and Ho¨llt V. (2000). Morphine self-administration in-opioid receptordeficient mice. Naunyn-Schmiedeberg’s Arch Pharmacol 361: 584-589.

Becker A. Grecksch G. Kraus J. Loh H.H. Schroeder H. and Ho¨llt V. (2002). Rewarding effects of ethanol and cocaine in mu opioid receptor-deficient mice. Naunyn- Schmiedeberg’s Arch Pharmacol 365: 296-302.

Berrettini W.H. Ferraro T.N. Alexander R.C. Buchberg A.M. and Vogel W.H. (1994). Quantitative trait loci mapping of three loci controlling morphine preference using inbred mouse strains. Nat Genet 7: 54-58.

Bohn L.M. Xu F. Gainetdinov R.R. and Caron M.G. (2000). Potentiated opioid analgesia in norepinephrine transporter knock-out mice. J Neurosci 20: 9040-9045.

Bond C. LaForge K.S. Tian M. Melia D. Zhang S. Borg L. Gong J. Schluger J. Strong J.A. Leal S.M. et al. (1998). Single nucleotide polymorphism in the human mu opioid receptor gene alters-endorphin binding and activity: possible implications for opiate addiction. Proc Natl Acad Sci USA 95: 9608-9613. Bowen D.V. Smit A.L.C. and Kreek M.J. (1978).F ecal excretion of methadone and its metabolites in man: application of GC-MS. in Advances in Mass Spectrometry (Daly NR ed.) pp. 1634-1639, Heyden and SonS. PhiladelphiA. PA.

Brzezinski M.R. Abraham T.L. Stone C.L. Dean R.A. and Bosron W.F. (1994). Purification and characterization of a human liver cocaine carboxylesterase that catalyzes the production of benzoylecgonine and the formation of cocaethylene from alcohol and cocaine. Biochem Pharmacol 48: 1747-1755.

Carroll K.M. Fenton L.R. Ball S.A. Nich C. Frankforter T.L. Shi J. and Rounsaville B.J (2004). Efficacy of disulfiram and cognitive behavior therapy in cocaine-dependent outpatients. Arch Gen Psychiatry 61: 264-272.

Carroll K.M. Ziedonis D. O’Malley S.S. McCance-Katz E. Gordon L.T. and Rounsaville B.J. (1993). Pharmacological interventions for abusers of alcohol and cocaine: a pilot study of disulfiram versus naltrexone. Am J Addict 2: 77-79.

Chen A.C.H. LaForge K.S. Ho A. McHugh P.F. Bell K. Schluger R.P. Leal S.M. and Kreek M.J. (2002). A potentially functional polymorphism in the promoter region of prodynorphin gene may be associated with protection against cocaine dependence or abuse. Am J Med Genet 114: 429-435.

Comings D.E. Blake H. Dietz G. Gade-Andavolu R. Legro R.S. Saucier G. Johnson P. Verde R. and MacMurray J.P. (1999). The proenkephalin gene (PENK) and opioid dependence. Neuroreport 10: 1133-1135.

Crowley J.J. Oslin D.W. Patkar A.A. Gottheil E. DeMaria P.A.Jr. O’Brien C.P. Berrettini W.H. and Grice D.E. (2003). A genetic association study of the mu opioid receptor and severe opioid dependence. Psychiatr Genet 13: 169-173. Dai D. Tang J. Rose R. Hodgson E. Bienstock R.J. Mohrenweiser H.W. and Goldstein J.A. (2001). Identification of variants of CYP3A4 and characterization of their abilities to metabolize testosterone and chlorpyrifos. J Pharmacol Exp Ther 299: 825-831.

Dean R.A. Christian C.D. Sample R.H. and Bosron W.F. (1991). Human liver cocaine esterases: ethanol-mediated formation of ethylcocaine. FASEB J 5: 2735-2739.

Dole V.P., Nyswander M.E. and Kreek M.J. (1966). Narcotic blockade. Arch Intern Med 118: 304-309.

Duguay Y., Skorpen F. and Guillemette C. (2004).Anovel functional polymorphism in the uridine diphosphate-glucuronsyltransferase 2B7 promoter with significant impact on promoter activity. Clin Pharmacol Ther 75: 223-233.

Eap C.B., Broly F., Mino A., Ha¨mmig R., De´glon J.J. Uehlinger C., Meili D., Chevalley AF., Bertschy G., Zullino D., et al. (2001). Cytochrome P450 2D6 genotype and methadone steady-state concentrations. J Clin Psychopharmacol 21: 229-234.

Eiselt R., Domanski T.L., Zibat A., Mueller R., Presecan-Siedel E., Hustert E., Zanger U.M., Brockmoller J., Klenk H.P., Meyer U.A., et al. (2001). Identification and functional characterization of eight CYP3A4 protein variants. Pharmacogenetics 11: 447-458.

Fernandes L.C., Kilicarlsan T., Kaplan H.L., Tyndale R.F., Sellers E.M. and Romach M.K., (2002). Treatment of codeine dependence with inhibitors of cytochrome P450 2D6. J Clin Psychopharmacol 22: 326-329.

Holthe M., Klepstad P., Idle J.R., Kaasa S., Krokan H.E. and Skorpen F. (2003). Sequence variations in the UDP-glucuronsyltransferase 2B7 (UGT2B7) gene: identification of 10 novel single nucleotide polymorphisms (SNPs) and analysis of their relevance to morphine glucuronidation in cancer patients. Pharmacogenomics J 3: 17-26.

Holthe M., Klepstad P., Zahlsen K., Borchgrevink P.C., Hagen L., Dale O., Kaasa S., Krokan H.E. and Skorpen F. (2002). Morphine glucuronide- to-morphine plasma ratios are unaffected by the UGT2B7 H268Y and UGT1A1*28 polymorphisms in cancer patients on chronic morphine therapy. Eur J Clin Pharmacol 58: 353-356.

Howard L.A., Sellers E.M. and Tyndale R.F. (2002). The role of pharmacogeneticallyvariable cytochrome P450 enzymes in drug abuse and dependence. Pharmacogenomics 3: 185-199.

Johnson R.E., Chutuape M.A., Strain E.C., Walsh S.L., Stitzer M.L. and Bigelow G.E. (2000). A comparison of levomethadyl acetatE. buprenorphinE. and methadone for opioid dependence. N Engl J Med 343: 1290-1297.

Kalow W. and Genest K. (1957). A method for the detection of atypical forms of human serum cholinesterase; determination of dibucaine numbers. Can J Med Sci 35: 339-346.

Kalow W. and Staron N. (1957). On distribution and inheritance of atypical forms of human serum cholinesterasE. as indicated by dibucaine numbers. Can J Biochem Physiol 35: 1305-1320.

Kamendulis L.M., Brzezinski M.R., Pindel E.V., Bosron W.F. and Dean R.A. (1996). Metabolism of cocaine and heroin is catalyzed by the same human liver carboxylesterases. J Pharmacol Exp Ther 279: 713-717.

Kathiramalainathan K., Kaplan H.L., Romach M.K., Busto U.E., Li N.Y., Tyndale R.F. and Sellers E.M. (2000). Inhibition of cytochrome P450 2D6 modifies codeine abuse liability. J Clin Psychopharmacol 20: 435-444.

Kitanaka N., Sora I., Kinsey S., Zeng Z. and Uhl G.R. (1998). No heroin or morphine 6 beta-glucuronide analgesia in -opioid receptor knockout mice. Eur J Pharmacol 355: R1-R3.

Kling M.A., Carson R.E., Borg L., Zametkin A., Matochik J.A., Schluger J., Herscovitch P., Rice K.C., Ho A., Eckelman W.C. et al. (2000). Opioid receptor imaging with PET and [18F]cyclofoxy in long-term methadone-treated former heroin addicts. J Pharmacol Exp Ther 295: 1070-1076.

Laizure S.C., Mandrell T., Gades N.M. and Parker R.B. (2003). Cocaethylene metabolism and interaction with cocaine and ethanol: role of carboxylesterases. Drug Metab Dispos 31: 16-20.

Lawford B.R., Young R.M., Noble E.P., Sargent J., Rowell J., Shadforth S., Zhang X. and Ritchie T. (2000). The D(2) dopamine receptor A(1) allele and opioid dependence: association with heroin use and response to methadone treatment. Am J Med Genet 96: 592-598.

Li S., Zhu J., Chen C., Chen Y.-W., Deriel J.K., Ashby B. and Liu-Chen L.-Y. (1993). Molecular cloning and expression of a rat opioid receptor. Biochem J 295: 629- 633.

Li T., Liu X., Zhao J., Hu X., Ball. DM., Loh el-W., Sham P.C. and Collier D.A. (2002). Allelic association analysis of the dopamine D2, D3, 5-HT2A. and GABA(A)gamma2 receptors and serotonin transporter genes with heroin abuse in Chinese subjects. Am J Med Genet 114: 329-335.

Li T., Liu X., Zhu ZH., Zhao J., Hu X., Sham P.C. and Collier D.A. (2000). Association analysis of polymorphisms in the opioid gene and heroin abuse in Chinese subjects. Addict Biol 5: 181-186.

Lima M.S., Soares B.G., Reisser A.A. and Farrell M. (2002). Pharmacological treatment of cocaine dependence: a systematic review. Addiction 97: 931- 949.

Lin Z. and Uhl G.R. (2003). Human dopamine transporter gene variation: effects of protein coding variants V55A and V382A on expression and uptake activities. Pharmacogenomics J 3: 159-168.

Ling W., Shoptaw S. and Majewska D. (1998). Baclofen as a cocaine anti- craving medication: a preliminary clinical study. Neuropsychopharmacology 18: 403-404.

Lockridge O., Mottershaw-Jackson N., Eckerson H.W. and La D.u. B.N. (1980). Hydrolysis of diacetylmorphine (heroin) by human serum cholinesterase. J Pharmacol Exp Ther 215: 1-8.

Loh H.H., Liu H.-C., Cavalli A., Yang W., Chen Y.-F. and Wei L.-N. (1998). opioid receptor knockout in mice: effects on ligand-induced analgesia and morphine lethality. Mol Brain Res 54: 321-326.

Lo¨tsch J., Skarke C., Gro¨sch S., Darimont J., Schmidt H. and Geisslinger G. (2002). The polymorphism A118G of the human -opioid receptor gene decreases the pupil constrictory effect of morphine-6-glucuronide but not that of morphine. Pharmacogenetics 12: 3-9.

Nath R.P., Upton R.A. and Everhart E.T. (1999). Buprenorphine pharmacokinetics: relative bioavailability of sublingual tablet and liquid formulations. J Clin Pharmacol 39: 619-623.

Romach M.K., Otton S.V., Somer G., Tyndale R.F. and Sellers E.M. (2000). Cytochrome P450 2D6 and treatment of codeine dependence. J Clin Psychopharmacol 20: 43-45.

Sawyer M.B., Innocenti F., Das S., Cheng C., Ramirez J., Pantle-Fisher FH., Wright C., Badner J., Pei D., Boyett J.M. et al. (2003). A pharmacogenetic study of uridine diphosphate-glucuronosyltransferase 2B7 in patients receiving morphine. Clin Pharmacol Ther 73: 566-574.

Sindrup S.H., Brøsen K., Bjerring P., Arendt-Nielsen L., Larsen U., Angelo H.R. and Gram L.F. (1991). Codeine increases pain thresholds to copper vapor laser stimuli in extensive but not poor metabolizers of sparteine. Clin Pharmacol Ther 49: 686- 693.

Sindrup S.H., Poulsen L., Brøsen K., Arendt-Nielsen L. and Gram L.F. (1993). Are poor metabolizers of sparteine/debrisoquine less pain tolerant than extensive metabolizers? Pain 53: 335-349.

Sivam S.P. (1989). Cocaine selectively increases striatonigral dynorphin levels by a dopaminergic mechanism. J Pharmacol Exp Ther 250: 818-824.

Skarke C., Darimont J., Schmidt H., Geisslinger G. and Lo¨tsch J. (2003a). Analgesic effects of morphine and morphine-6-glucuronide in a transcutaneous electrical pain model in healthy volunteers. Clin Pharmacol Ther 73: 107-121. Spangler R., Ho A., Zhou Y., Maggos C., Yuferov V. and Kreek M.J. (1993a). Regulation of kappa opioid receptor mRNA in the rat brain by “binge” pattern cocaine administration and correlation with preprodynorphin mRNA. Mol Brain Res 38: 71-76.

Tyndale R.F., Droll K.P. and Sellers E.M. (1997). Genetically deficient CYP2D6 metabolism provides protection against oral opiate dependence. Pharmacogenetics 7: 375-379.

Xie W., Altamirano C.V., Bartels C.F., Speirs R.J., Cashman J.R. and Lockridge O. (1999). An improved cocaine hydrolase: the A328Y mutant of human butyrylcholinesterase is 4-fold more efficient. Mol Pharmacol 55: 83-91.

Xu K., Liu X.H., Nagarajan S. Gu X.Y. and Goldman D (2002) Relationship of the -opioid receptor gene to heroin abuse in a large Chinese case/control sample. Am J Med Genet 110: 45-50.

Yakovlev A.G., Krueger K.E. and Faden AI. (1995). Structure and expression of a rat kappa opioid receptor gene. J Biol Chem 270: 6421-6424.

Yasuda K., Raynor K., Kong H., Breder C.D., Takeda J., Reisine T. and Bell GI. (1993). Cloning and functional comparison of and opioid receptors from mouse brain. Proc Natl Acad Sci USA 90: 6736-6740.

Yuferov V., Fussell D., LaForge K.S., Nielsen D.A., Gordon D., Ho A., Leal S.M., Ott J. and Kreek M.J. (2004). Redefinition of the human kappa opioid receptor gene (OPRK1) structure and association of haplotypes with opiate addiction. Pharmacogenetics 14: 793-804.

Yuferov V., Zhou Y., Spangler R., Maggos C.E., Ho A. and Kreek M.J. (1999). Acute “binge” cocaine increases -opioid receptor mRNA levels in areas of rat mesolimbic mesocortical dopamine system. Brain Res Bull 48: 109-112.

Zhu J., Chen C., Xue J.-C., Kunapuli S., DeRiel J.K. and Lui-Chen L.-Y. (1995). Cloning of a human opioid receptor from the brain. Life Sci 56: PL201- PL207.

Zubieta J., Gorelick D.A., Stauffer R., Ravert H.T., Dannals R.F. and Frost J.J. (1996). Increased mu opioid receptor binding detected by PET in cocaine- dependent men is associated with cocaine craving. Nat Med 2: 1225-1229

Descargas

Los datos de descargas todavía no están disponibles.
Sistema OJS - Metabiblioteca |